Abstract Cancer stem cells (CSCs) play a crucial role in the progression of tumors, resistance to immunotherapy, and the development of thyroid carcinoma (THCA). However, the precise mechanisms involved remain poorly understood. In this study, 26 mRNAs linked to cancer stemness were identified through an analysis of 13 publicly available THCA transcriptomic datasets alongside a CRISPR dataset for thyroid cancer cell lines. Notably, we revealed an inverse relationship between the stemness of cancer cells and the efficacy of immunotherapy. Utilizing multiple machine learning approaches, we created and confirmed a model for tumor stemness cells (TSC), which included genes associated with cancer stemness. This model successfully forecasted patient outcomes and responses to treatment with Cytotoxic T-Lymphocyte Associated Protein 4 (CTLA4) immune checkpoint inhibitors (ICIs) across different types of cancer. Additionally, chromosomal amplifications in regions 1q and 8p were identified as intrinsic contributors to the onset of thyroid cancer. Specifically, based on single cell dataset, the amplification of CKS1B on chromosome 1q was revealed to advance the progression of thyroid cancer cells by boosting their proliferation. Furthermore, experimental results revealed that the heightened expression of CKS1B considerably enhanced the proliferation and invasion capabilities of THCA, suggesting it could serve as a possible therapeutic target for this condition. In summary, our study sheds light on the relationship between the traits of cancer stem cells and the efficacy of immunotherapy, providing a novel model for predicting the prognosis and individual responses to immunotherapy in patients with THCA. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-02813-8. Keywords: Cancer stemness, Immunotherapy, Thyroid cancer, Single cell, CKS1B Introduction Thyroid cancer, which arises from the cells of the thyroid gland, is a prevalent malignancy and the most frequently diagnosed malignant tumor within the endocrine system, with an annual incidence increasing by an average of 4.5% [[40]1]. Despite advancements in diagnostic techniques and therapeutic strategies, the ability to rapidly detect and precisely predict patient prognosis remains limited [[41]2]. Focusing on the inhibitory interactions that occur between immune checkpoint receptors and their corresponding ligands, including the programmed death 1 (PD-1) pathway, has transformed the approach to treating advanced cancers through the use of immune checkpoint inhibitors. The integration of immunotherapy has transformed the approach to cancer care and enhanced patient survival rates. However, due to the genetic diversity of tumors, current biomarkers are not adequate for accurately categorizing patients for optimal treatment outcomes [[42]3]. Certain patients might not react positively and may encounter adverse effects, highlighting the significance of recognizing strong markers and enhancing combinations of biomarkers. Precisely identifying patients who will benefit from immunotherapy could expand the pool of eligible candidates for this treatment, potentially lessening the impact of chemotherapy side effects in individuals with thyroid cancer. CSCs exhibit a pronounced capacity for self-renewal, display considerable heterogeneity along with metastatic potential, and contribute significantly to tumor initiation, progression, and dissemination [[43]4, [44]5]. The diversity of cellular components and the immune-suppressive milieu within the tumor serve as distinct yet interrelated factors driving tumor progression and fostering treatment resistance [[45]6]. Prior investigations have demonstrated extensive negative associations between cancer stem cell traits and the immune system's response to cancer therapy. This phenomenon was noted even though tumors with high stemness exhibited elevated mutation rates, cancer-specific antigen expression, and intratumor heterogeneity across 21 different solid cancer types [[46]7]. Recent study revealed a negative correlation between tumor stemness and the therapeutic outcomes of ICIs in various cancers. Their research introduced a robust predictive model to investigate how cancer stemness influences immunotherapy responsiveness [[47]8]. Despite these advancements, the specific impact of tumor stemness on ICI efficacy in thyroid cancer remains underexplored. In this research, we effectively employed the robust mRNA-based stemness index (mRNAsi) [[48]9] to pinpoint 26 mRNAs associated with cancer stemness. Through comprehensive analysis of transcriptomic profiles, single-cell data, and CRISPR-based cell line studies, our findings demonstrated an inverse association between cancer stemness and the efficacy of immunotherapy. Subsequently, we created and validated a scoring model for TSC, designed to forecast prognosis and responses to immunotherapy in individuals diagnosed with thyroid cancer. Additionally, we discovered that the overexpression of CKS1B, a crucial gene associated with cancer stemness, can stimulate the growth of thyroid cancer cells. This gene could potentially act as a valuable biomarker for predicting the cellular origins of malignant thyroid cells, supported by experiments conducted with cell lines. Our research offers fresh insights into the connection between CSCs and immunotherapy in the context of THCA. Furthermore, it introduces a well-performed predicting model for categorizing THCA patients based on their immune responses and survival outcomes. Methods Public data collection The dataset concerning thyroid cancer with survival data from The Cancer Genome Atlas (TCGA) was acquired from the UCSC Xena database [[49]10]. In addition, 12 GEO cohorts were studied to focus on thyroid cancer ([50]GSE104005, [51]GSE196264, [52]GSE29265, [53]GSE32662, [54]GSE35570, [55]GSE53157, [56]GSE5364, [57]GSE60542, [58]GSE65074, [59]GSE65144, [60]GSE76039, and [61]GSE82208) to examine mRNAs associated with cancer stemness. Furthermore, the gene expression profiles of a single-cell dataset for thyroid cancer with accession number [62]GSE184362 [[63]11] were collected from the GEO database to explore mRNAs linked to intrinsic stemness in cancer cells. To authenticate the precision of the model in forecasting the response to CTLA4 immunotherapy, we collected diverse datasets from cohorts treated with anti-CTLA4. These included the cohort by Riaz N [[64]12] ([65]GSE91061: melanoma patients administered with anti-CTLA4 and PD-1) and the cohort by Necchi [[66]13] (IMvigor210: individuals with advanced or metastatic urothelial carcinoma treated with Atezolizumab) obtained via the "IMvigor210CoreBiologies" R package. We amassed gene expression and clinical data from these cohorts. To explore the relationship between the stemness properties of cancer cells and their response to immunotherapy, a cohort of melanoma patients treated with ICIs was analyzed. This analysis incorporated single-cell RNA sequencing data and ICI response information, accessible from GEO under accession number [67]GSE115978 [[68]14]. Experimental data and material Human thyroid cancer cell line (KHM-5M) was obtained from the Cell Bank of the Committee for Conservation of Typical Cultures at the Chinese Academy of Sciences. These cell lines were cultured in Dulbecco's Modified Eagle Medium (DMEM) (HyClone, Logan, UT, USA), supplemented with 10% fetal bovine serum, 100 IU/mL penicillin, and 100 IU/mL streptomycin (Gibco, New York, USA). The siRNA targeting CSK1B was synthesized by Biotend Co., Ltd. Cells were transfected with 50 nM of siRNA for 24 h utilizing the jetPRIME transfection kit from Polyplus-transfection (Strasbourg, France). The Western blotting protocol was performed as follows: Cultured cells were washed with ice-cold PBS, and total protein lysates were extracted at 4 °C using RIPA lysis buffer (Beyotime, Shanghai, China) supplemented with 1% protease inhibitor cocktail (MedChemExpress, New Jersey, USA). After centrifugation at 12,000 g for 20 min at 4 °C, the supernatant was collected and mixed with loading buffer. The samples were then separated by 10% SDS-PAGE and transferred to a PVDF membrane. The membrane was blocked with 5% skim milk for 2 h at room temperature and incubated overnight at 4 °C with primary antibodies. After washing with Tris Buffered Saline, the membrane was incubated with secondary antibodies. Protein detection was performed using enhanced chemiluminescence reagents (Beyotime, Shanghai, China). The following antibodies were used for Western blot analysis: CKS1B (ab72639, Abcam, Waltham, Massachusetts, USA) and GAPDH (60004-1-Ig, Proteintech, Wuhan, China). To evaluate clonogenic potential, 1000 to 2000 cells were seeded into each well of a 6-well plate and cultured for 7 to 10 days. Colonies containing more than 50 cells were fixed with 0.2% crystal violet solution for 30 min. After washing three times with PBS, the colonies were photographed and quantified. Cells were evenly plated in a 6-well plate. Upon reaching confluence, a straight scratch was created using a 10-μL pipette tip guided by a ruler. After PBS washing, the cells were maintained in serum-free medium. Incubation was carried out at 37 °C with 5% CO2, and images were captured at 0 and 24 h post-scratch. To assess cell migration, 3–5 × 10^4 cells were suspended in 200 μL of serum-free medium and added to the upper chamber of Transwell plates (BD Biosciences, Bedford, MA, USA). The lower chamber was filled with 600 μL of medium containing 10% FBS. After 24 h of incubation at 37 °C, the cells were fixed with 4% paraformaldehyde for 30 min and stained with 0.25% crystal violet for an additional 30 min. Non-migrated cells on the upper side of the membrane were removed, and migrated cells on the lower side were imaged and counted. Statistical analysis CRISPR screening of thyroid carcinoma cells was conducted using the DepMap portal ([69]https://depmap.org/portal/download). The CERES algorithm was employed to calculate dependency scores for approximately 17,000 genes. Genes identified as essential exhibited a CERES score lower than −1 in 75% of the 21 THCA cell lines analyzed [[70]15]. A novel pipeline was developed to establish a predictive model for cancer stem cells, as depicted in Fig. [71]1. Firstly, the calculation of mRNAsi was conducted on 12 GEO datasets as well as the TCGA-THCA dataset based on the pipeline described in previous research [[72]16]. Utilizing a predefined reference gene signature containing specific stem cell-associated genes, each with assigned weight coefficients [[73]16], we extracted the common genes between this signature and our datasets. Stemness scores were computed by calculating the Spearman correlation between each sample's gene expression profile and the reference stem cell gene signature, which quantifies the similarity of gene expression patterns to the canonical stem cell gene set. To facilitate interpretability and comparative analysis, we linearly transformed the correlation scores to a standardized 0–1 range, where higher scores indicate greater stem cell-like characteristics. Subsequently, the connection between overall mRNA and mRNAsi was evaluated, and mRNAs showing noteworthy positive correlations in a minimum of seven out of thirteen sets (Cor > 0.2 and P < 0.05) were recognized as mRNAsi-related mRNAs, leading to the identification of 135 mRNAsi-linked mRNAs. Fig. 1. [74]Fig. 1 [75]Open in a new tab Identification of key mRNAs linked to cancer stemness and their association with immunotherapy response. A Thyroid cancer cohorts used to identify mRNAs correlated with cancer stemness. B Schematic representation of the CRISPR screening process for mRNAs involved in thyroid cancer progression. C Venn diagram depicting the overlap between mRNAs associated with cancer stemness and those linked to thyroid cancer prognosis. D Box plot comparing the normalized expression levels of 26 stemness-related mRNAs in normal versus tumor thyroid tissues. E KEGG pathway enrichment analysis of mRNAs related to cancer stemness. F tSNE plot classifying malignant cells based on response phenotypes. Red points represent malignant cells from NR patients, and dark blue points represent malignant cells from TN patients. G tSNE plot visualizing AUCell scores for cancer stem cell-related gene sets in malignant cells, with red representing high stemness and blue indicating low stemness. H Box plot illustrating the distribution of AUCell scores across response phenotypes (NR vs. TN) in the SKCM cohort (Wilcoxon test; *P < 0.05; **P < 0.01; ***P < 0.001) As a result of the scarcity of clinical survival data on thyroid cancer, the TCGA-THCA dataset was split into two halves, Cohort1 and Cohort2, while all samples were merged into Cohort3. We utilized Cohort1 as the training set to develop machine learning model, then validated the model's accuracy using both Cohort2 and Cohort3. Afterwards, the effectiveness of the stemness model for cancer was created and assessed. To develop the cancer stemness model, various machine learning algorithms were utilized, such as random forest (RSF) [[76]17], elastic net (Enet) [[77]18], gradient boosting (GBM) [[78]19], survival-SVM [[79]20], ridge regression [[80]21], Stepcox [[81]22], plsRcox [[82]23], CoxBoost [[83]24], lasso [[84]25], and SuperPC [[85]26]. Specifically, through the input of the expression matrix of 26 selected cancer stemness related genes, the corresponding machine learning algorithm was used to predict the survival time and survival state of patients, and finally the score for each patient was calculated. To evaluate the effectiveness of PD-1/CTLA4 immunotherapy, we initially computed tumor immune dysfunction and exclusion (TIDE) scores using expression data from thyroid cancer patients. These expression profiles were subsequently analyzed on the TIDE database platform [[86]27] ([87]http://tide.dfci.harvard.edu/) to predict patient outcomes by uploading the expression matrix into [88]http://tide.dfci.harvard.edu/ and ‘cancer type’ section selected ‘other’ and ‘Previous immunotherapy’ section selected ‘no’. We prepared input files for SubMap analysis through a meticulous data processing workflow. Initially, we utilized raw expression data from the original dataset, applying rigorous filtering to remove low-expressed genes (defined as those with values less than 1 in over 90% of samples). To standardize the gene expression data, we identified the intersection of gene lists between different datasets and performed log2 transformation by adding 1 to each value (log2(x + 1)) to normalize the expression scale and mitigate potential numerical issues. Sample classification was conducted by categorizing samples into two distinct clusters: low-TSC and high-TSC. Each cluster was assigned a numeric rank to facilitate computational analysis, with TSC-low designated as rank 1 and TSC-high as rank 2. R function, generateInputFileForSubMap(), was used to systematically generate two critical files required for SubMap analysis: SubMap.gct and SubMap.cls. The SubMap.gct file encapsulates the log2-transformed gene expression matrix, preserving the molecular details of each sample, while the SubMap.cls file contains the corresponding sample classification information with their respective numeric ranks. This approach ensures that the files meet the stringent input requirements of the SubMap algorithm, enabling precise comparative analysis of gene expression profiles across different immunological clusters. Subsequently, the submap algorithm on the GenePattern portal was utilized to compare response probabilities between low- and high-TSC groups. In single cell analysis, the expression profiles for all samples were combined through the Merge() function in R (version 3.6.1). Next, library size normalization took place using the NormalizeData() function within Seurat [[89]28], resulting in normalized counts that were then log-transformed. The 2000 most highly variable genes were identified using the FindVariableFeatures() function. Principal components (PCs) were calculated from the expression profiles of these top 2000 variable genes, focusing on the top 30 components. For cell clustering, the FindNeighbors() and FindClusters() functions in Seurat were employed. When suitable, visualization was carried out using the RunUMAP() functions. A 2D Uniform Manifold Approximation and Projection (UMAP) algorithm facilitated the visualization of cells using the RunUMAP() and DimPlot() functions. Marker genes for each cluster were identified through the FindAllMarkers() function in Seurat. For any specific cluster, the FindAllMarkers() function revealed positive markers by comparing them with all other clusters. Clinical characteristics between high and low TSC score groups were compared using the Wilcoxon rank-sum test. KEGG enrichment is carried out through the enrichKEGG function in the clusterProfiler package [[90]29]. Additionally, GSEA enrichment of HALLMARK pathways was performed using the GSEA function from the clusterProfiler package. The HALLMARK dataset was obtained from the MisgDB database ([91]https://www.gsea-msigdb.org/gsea/index.jsp). The logFC values of input genes were calculated using the FindMarkers function based on the comparison between CKS1B+ malignant and CKS1B- malignant cells. Differences in immunotherapy response rates between the two groups were analyzed using the Chi-square test. Pearson’s correlation coefficient was calculated to examine the association between mRNA expression and mRNAsi. The relationship between TSC, CKS1B, and patient survival was evaluated using Kaplan-Meier survival analysis, with statistical significance assessed by the log-rank test. Time-dependent receiver operating characteristic (ROC) curves, generated using the 'pROC' [[92]30] R package, were employed to evaluate the prognostic and immunotherapy predictive value of TSC. The C-index was calculated based on the model-predicted scores, utilizing the coxph function to evaluate patients' survival time and survival status. Furthermore, to evaluate whether the TSC model could serve as an independent prognostic factor, we utilized the coxph function to perform multivariate Cox regression analysis. We calculated the impact of various clinical features on patient prognosis. Specifically, features with P < 0.05 were considered to have an independent predictive capability for patient survival, unaffected by the influence of other clinical features. Patients were stratified into groups based on the optimal cutoff value determined by the 'survminer' R package. A significance level of P or adjP < 0.05 was considered statistically significant. Results Cancer stemness is negatively associated with immunotherapy benefits A comprehensive analysis was conducted to investigate the association between cancer stemness and immunotherapy efficacy. Thirteen transcriptomic datasets for THCA were retrieved from the GEO and TCGA databases to identify mRNAs linked to cancer stemness, with mRNAsi values calculated for each patient (Fig. [93]1A). Using Pearson’s correlation coefficient, mRNAs exhibiting a significant positive correlation with mRNAsi in most cohorts (over 50%; 7 out of 13; Cor > 0.2 and P < 0.05) were selected. A total of 135 mRNAs were then singled out as being related to cancer stemness, signifying the stemness level within the tumor cells. Furthermore, a thorough examination of loss-of-function screens based on CRISPR technology was conducted globally utilizing the DepMap database to pinpoint significant candidate genes linked to THCA malignancy. We finally identified 647 genes critical for the survival of 21 THCA cell lines (CERES score < − 1 in 75% of malignant THCA cells). Among these, 26 mRNAs were selected based on the overlap between THCA mRNAsi-associated mRNAs and those identified through CRISPR screening (Fig. [94]1C). Consistent with above results, the majority of the identified genes displayed significant overexpression in tumor tissues compared to adjacent tissues, providing evidence of their role in tumor progression (Fig. [95]1D). Additionally, the KEGG enrichment demonstrated that these genes are mainly engaged in biological processes like the cell cycle and DNA repair, which further reinforces their connection to the stemness of tumor stem cells (Fig. [96]1E). All selected genes were prioritized for further investigation. To assess the impact of cancer stemness-related genes on immunotherapy efficacy, we analyzed a published single-cell dataset from melanoma patients treated with immune checkpoint inhibitors (ICIs), focusing on the association between cancer stemness and ICI response. After excluding samples lacking malignant cells, we concentrated on 23 patients, including 10 non-responders (NR) and 13 treatment-naïve (TN) individuals. Using AUCell [[97]31], based on the 26 selected genes related to cancer stemness, we calculated the scores of cancer stemness-related genes in each cell using the AUCell_calcAUC method. We observed that malignant cells from the NR subgroup displayed significantly higher stemness levels (P < 0.001, Fig. [98]1H), indicating an inverse correlation between cancer stemness and ICI treatment outcomes. Taken together, our results indicated that these 26 mRNAs above selected can accurately characterized cancer stemness, and the stemness of cancer is negatively correlated with immunotherapy benefits. Establishment of cancer stemness score based on THCA RNA-seq dataset To advance the development of a prediction model aimed at assessing the response to immunotherapy based on mRNAs associated with cancer stemness, a variety of machine learning algorithms were employed. These predictors were used to calculate scores for each sample across three cohorts. The evaluation of performance involved determining the average C-index for every algorithm, which showed that the majority of predictors yielded a high average C-index (Fig. [99]2A). Among the algorithms, the RSF approach demonstrated the greatest accuracy, achieving an average C-index of 0.993 (Fig. [100]2A), and was designated as the TSC score. Subsequently, patients were stratified according to the optimal cutoff determined by the 'survminer' R package, revealing that individuals with higher TSC scores had significantly reduced overall survival (OS) compared to those with lower TSC scores in cohorts 1–3 (Figs. [101]2B, S1A, B). Additionally, our results demonstrated that TSC is a reliable prognostic indicator, as evidenced by area under the curve (AUC) values. The AUC analysis highlighted strong predictive accuracy, with an average AUC exceeding 0.98 across the three cohorts analyzed (Fig. [102]2C). To enhance the clinical applicability of our model, we investigated whether TSC could function independently of other clinical variables. A comprehensive multi-variable cox analysis indicated that TSC could act as an autonomous prognostic factor for assessing THCA prognosis (Fig. [103]2D). Recent progress in next-generation sequencing and large-scale data mining has enabled a comprehensive exploration and refinement of gene expression-based prognostic markers. To rigorously evaluate the performance of TSC compared to other markers, we systematically reviewed 79 models developed over the past decade (Table S1). Notably, TSC outperformed all other models in predicting survival outcomes across three independent THCA cohorts, achieving an average AUC greater than 0.98 in these cohorts (Fig. [104]2E). Fig. 2. [105]Fig. 2 [106]Open in a new tab Construction of a cancer stemness model using diverse machine learning approaches. A C-index for each algorithm across three independent cohorts, where the color of each grid represents the magnitude of the C-index obtained by different algorithms/algorithm combinations, and the bar graph on the right represents the average C-index across the three cohorts. B Kaplan-Meier survival curves comparing overall survival in TCGA-THCA patients stratified by high and low TSC scores in cohort3. C AUC values of the TSC model for predicting clinical status in three THCA cohorts. D Multivariate Cox regression analysis evaluating the independent prognostic significance of the TSC score in TCGA-THCA samples. E ROC values demonstrating the predictive performance of the TSC model compared to 79 other models for clinical status in three THCA cohorts In summary, the results demonstrated that the TSC model is a reliable prognostic model for THCA, exhibiting substantially greater predictive accuracy than other existing models. Tumor stemness cell score as a potential predictor for CTLA4 immunotherapy outcomes Given the inverse correlation between cancer stemness and immunotherapy efficacy, we investigated the potential of TSC as a predictive biomarker for immunotherapy outcomes in THCA patients. Using the submap algorithm on the GenePattern platform, we assessed the likelihood of immunotherapy response in patients stratified by high or low TSC levels. Notably, the low TSC group showed a significantly improved response to CTLA4 immunotherapy (P = 0.072, Bonferroni corrected P < 0.01, Fig. [107]3A), underscoring the utility of the TSC model in predicting CTLA4 immunotherapy benefits. Fig. 3. [108]Fig. 3 [109]Open in a new tab Evaluation of the cancer stemness model as a predictor of immunotherapy efficacy. A Predicted response rates to immune checkpoint inhibitors (CTLA4 and PD1) across TSC subgroups (R: Response, NR: No Response). B, C Kaplan-Meier survival curves depicting overall survival in patients receiving CTLA4 immune checkpoint inhibitor (ICI) therapy in metastatic urothelial carcinoma (IMvigor210; B) and melanoma ([110]GSE91061; C). D Immunotherapy response rates stratified by TSC groups, with responses classified as complete (CR) or partial (PR), and non-responses as progressive (PD) or stable disease (SD). E, F Receiver operating characteristic (ROC) curves illustrating the TSC model's ability to predict response status in CTLA4 ICI-treated cohorts To further confirm the ability of TSC to predict the therapeutic effectiveness of CTLA4 ICIs, we gathered a variety of datasets associated with CTLA4 ICIs. Our findings consistently revealed that patients exhibiting lower TSC scores experienced notably improved overall survival rates after undergoing CTLA4 immunotherapy when compared to those presenting higher TSC scores (Fig. [111]3B and C). This indicates that elevated TSC scores might hinder the advantages of CTLA4 immunotherapy. Patients with elevated TSC scores demonstrated less favorable responses to the therapy, with merely 48% exhibiting a positive reaction (Fig. [112]3D). Significantly, our analysis demonstrated that TSC reliably predicts response to CTLA4-ICI therapy, as supported by high area under the curve (AUC) values. The AUC analysis revealed strong predictive performance, with values of 0.996 at 1 year, 0.989 at 2 years, and 0.989 at 3 years in the IMvigor210 cohort, and 0.900 at 1 year, 0.975 at 2 years, and 0.854 at 3 years in the SKCM (melanoma) cohort (Fig. [113]3E and F). To summarize, the TSC model serves as a dependable predictor for projecting the outlook of THCA patients and acts as an efficient biomarker for anticipating patient response to immunotherapy. The amplification of CKS1B is associated with the proliferation of malignant cells in THCA To investigate the clonal composition and cellular heterogeneity of thyroid cancer, we collected single-cell RNA dataset of thyroid carcinoma patients. Cells expressing fewer than 200 genes or exhibiting mitochondrial gene content exceeding 20% were filtered out. Following quality control, the retained cells were categorized into eight major cell types using established biomarkers. Figure [114]4A and B illustrate distinct clusters, encompassing B cells, endothelial cells, epithelial cells, fibroblasts, myeloid cells, plasma cells, T & NK cells, and dendritic cells. Fig. 4. [115]Fig. 4 [116]Open in a new tab Exploring core endogenous heterogeneity driven cancer stemness associated gene influencing prognosis of thyroid cancer. A UMAP plot depicting cell types annotated in thyroid cancer dataset with the accession number of [117]GSE184362. B Heatmap showing expression of each marker in each cell type. C Heatmap showing copy number variation of reference cells and epithelial cells. D Venn plot selected the cancer stemness associated mRNAs driven from thyroid malignant cell endogenous heterogeneity. E Box plot illustrating the normalized expression of CKS1B between normal and tumor tissues in thyroid cancer. F CKS1B expression in THCA tissues from human protein atlas. G Box plots comparing CKS1B expression between early (Stage I/II or T1/T2) and advanced (Stage III/IV or T3/T4) THCA groups. H GSEA enrichment of HALLMARK pathways between CKS1B- and CKS1B+ malignant cells Subsequently, we employed the inferCNV computational approach to examine both copy number variations (CNV) and clonal diversity within thyroid malignancy cells originating from epithelial cells (ECs). In the analysis, a total of 29,499 ECs from THCA tissues were examined, revealing that 21,052 of these cells displayed elevated CNV scores. This substantial number of cells exhibiting high CNV is indicative of malignancy, highlighting a significant characteristic of tumorigenesis in these instances (Fig. [118]4C). Moreover, particular amplifications were noted in chromosomal 8p and 1q, which emerged as significant driving variations associated with the pathogenesis of thyroid cancer. Notably, the amplification of the CKS1B gene located on chromosomal 1q was found to be linked with genes related to stemness (Fig. [119]4D). Previous research has established that the CKS1B gene has a pivotal role in promoting cell proliferation and invasion through the activation of the STAT3/AKT signaling pathway, particularly in cases of papillary thyroid carcinoma [[120]32]. This connection underscores the potential of CKS1B as a critical factor in the progression of thyroid malignancies. CKS1B expression was consistently found to be markedly increased in tumors when compared to the adjacent para-cancerous tissues in TCGA (Fig. [121]4E). Additionally, aligning with our bioinformatics findings, the protein levels of CKS1B were significantly higher in thyroid cancer tissues in contrast to normal thyroid samples (Fig. [122]4F). Moreover, our observations indicated that elevated CKS1B expression was particularly associated with the progression of tumor size and stage in TCGA (Fig. [123]4G). In conclusion, the upregulation of CKS1B may propel the advancement of malignant thyroid cells and could function as a valuable biomarker for predicting the cellular origins of THCA cancerous cells. CKS1B enhances thyroid malignant cells’ proliferation, migration, and invasion To investigate the impact of CKS1B on thyroid cancer cells, we categorized malignant tumor cells with an expression value above 1 as CKS1B+ malignant tumor cells, while those with an expression value of 0 were classified as CKS1B- malignant tumor cells based on CKS1B expression levels. Our HALLMARK enrichment analysis revealed that CKS1B is capable of activating cell cycle-related pathways such as the G2M checkpoint and E2F targets (Fig. [124]4H). To further validate the oncogenic role of CKS1B in thyroid cancer, we generated CKS1B knockdown model in KHM-5M cell line. Their efficacy was confirmed through western blot analyses at the protein level (Fig. [125]5A). Notably, CKS1B knockdown led to a marked inhibition of clonogenic capacity in KHM-5M cells (Fig. [126]5B), suggesting a role for CKS1B in promoting KHM-5M cell growth potential. Furthermore, in wound healing and Transwell assays, CKS1B knockdown significantly impaired the migratory capacity of KHM-5M cells (Fig. [127]5C), indicating an important role for CKS1B in promoting the metastatic potential of thyroid cancer. Fig. 5. [128]Fig. 5 [129]Open in a new tab CKS1B enhances thyroid cancer proliferation and migration in vitro. A Validation of CKS1B knockout in KHM-5M cells via Western blot analysis. B Clonal formation ability of CKS1B knockout KHM-5M cells. C Evaluation of migratory abilities in CKS1B knockout KHM-5M cells using wound healing and Transwell assays. Scale bar, 100 μm Discussion Immunotherapy, particularly immune checkpoint inhibitors, is now widely accepted as a second-line treatment for advanced cancers and is increasingly being approved for first-line use. Variability in tumors and their stem-like properties play crucial roles in how tumors evade the immune system and respond to immunotherapy. While significant research has been conducted on the relationship between cancer stem-like characteristics and anti-tumor immunity, direct evidence connecting tumor stemness to immune checkpoint inhibitor responses in thyroid cancer is presently lacking. Immunotherapy for thyroid cancer faces various challenges, with the main hurdle being the identification of suitable candidates for ICI treatment. In this research study, we employed an established stemness index [[130]9] to detect 28 cancer stemness-linked mRNAs in 13 thyroid cancer datasets. These gene sets were identified to participate in various important pathways including cell cycle, DNA replication, nucleotide excision repair, and the p53 signaling route. These pathways are crucial for regulating cancer proliferation [[131]33–[132]35]. Furthermore, our investigation revealed an inverse relationship between stemness and the outcomes of immune checkpoint inhibitors, supported by data from an ICI-treated single-cell cohort of SKCM [[133]14]. Moreover, several studies have indicated that patients with heightened cancer stemness experience deteriorated prognosis and heightened resistance to immunotherapy [[134]27, [135]36]. Subsequently, to assess the prognostic effect of tumor stemness genes in thyroid cancer patients, we utilized diverse machine learning algorithms to establish a prognostic model for survival and response to immunotherapy. The performance of this model was authenticated in three datasets, with the Random Survival Forest model being identified as the favored TSC model due to its improved reliability and precision. Analysis using the Kaplan-Meier method suggested that individuals with elevated TSC scores in THCA showed markedly poorer survival rates than those with lower TSC scores. Additionally, the AUC curve demonstrated an outstanding ability to predict outcomes, with the average AUC surpassing 0.98 across the three datasets. Additionally, our TSC score was validated as an autonomous predictor in multivariable Cox regression analysis. The TSC model showcased superior predictive performance for survival in contrast to conventional clinical and pathological characteristics, surpassing seventy-nine previously proposed models. The predictive value of cancer stemness for immunotherapy in various cancer types has been well-established, but its potential role in thyroid cancer remains largely unexplored [[136]8]. To assess the effectiveness of the cancer stemness index in forecasting the response to immunotherapy, we utilized the TIDE and Submap web server tools to analyze the immunotherapy response in thyroid cancer [[137]27]. The effectiveness of the TSC model in anticipating the reaction to CTLA4 immune checkpoint inhibitors, particularly in cases of thyroid cancer, is emphasized by our findings. These findings suggest that the TSC model may have broad utility in predicting CTLA4 ICI treatment responses across different cancer types. Then, a thorough analysis showed that TSC exhibited a high level of accuracy in forecasting CTLA4 ICIs responses across different datasets utilizing bulk RNA-Seq data, achieving an average AUC greater than 0.9. The presence of stemness is positively associated with increased intratumoral heterogeneity, potentially providing a protective mechanism for antigenic clones against elimination by the immune system [[138]7]. CNV in cancerous cells plays a crucial role in shaping tumor heterogeneity. Initial identification of inherent CNVs in malignant thyroid cells at the single-cell level indicated amplification of chromosomal 8p and 1q. Subsequent analysis linking mRNAsi and CNV profiles unveiled CKS1B as an endogenous mRNA linked to amplification-driven cancer stemness. As a protein-encoding gene, CKS1B associates with catalytic elements of cyclin-dependent kinases and enhances retinoblastoma cell proliferation, migration, invasion, and neovascularization via the MEK/ERK signaling cascade [[139]37]. Studies have earlier established CKS1B's utility as an immunotherapy response marker in pancreatic cancer patients [[140]38]. In line with these findings, our study confirms a notable upregulation of CKS1B in thyroid tumor tissue, correlating with tumor advancement. On a mechanistic level, overexpression of CKS1B triggers the G2M checkpoint and E2F target-related pathways, bolstering proliferation, invasion, and metastasis of thyroid cancer cells, presenting CKS1B as a potential therapeutic target for THCA. To summarize, based on single-cell dataset analysis from melanoma patients, our study suggests a potential inverse correlation between cancer stemness and immunotherapy response, which we applied to explore implications in thyroid cancer. Notably, we have developed a strong cancer stemness signature that can assess prognosis and potential benefits of immunotherapy, providing a useful tool for improving decision-making and monitoring strategies for individual patients with THCA. While we have shown the impressive predictive precision of the TSC in evaluating thyroid cancer prognosis and immunotherapy benefits, it is crucial to recognize some limitations in our research. The TSC model's ability to forecast immunotherapy outcomes for thyroid cancer is dependent on predictions from the submap and TIDE algorithm, and further validation of the TSC model using real thyroid cancer CTLA4 immunotherapy cohorts is required. Moreover, conducting studies with larger cohort data is vital to validate the performance of the TSC model in thyroid cancer. Supplementary Information [141]12672_2025_2813_MOESM1_ESM.tif^ (4.3MB, tif) Fig. S1 Validation of a cancer stemness model.A, B Kaplan-Meier survival curves comparing overall survival in TCGA-THCA patients stratified by high and low TSC scores in cohort1 (A) and cohort2 (B) Additional file1 (TIF 4417 kb) [142]Additional file2 (DOCX 92 kb)^ (92.6KB, docx) [143]Additional file3 (XLS 99 kb)^ (99KB, xls) Acknowledgments